{"id":795,"date":"2023-07-08T12:54:04","date_gmt":"2023-07-08T12:54:04","guid":{"rendered":"https:\/\/milkyeggs.com\/?p=795"},"modified":"2023-10-30T22:53:48","modified_gmt":"2023-10-30T22:53:48","slug":"lifespan-extension-separating-fact-from-fiction","status":"publish","type":"post","link":"https:\/\/milkyeggs.com\/biology\/lifespan-extension-separating-fact-from-fiction\/","title":{"rendered":"Lifespan extension: separating fact from fiction"},"content":{"rendered":"\n

Thinking about longevity practically is a tricky affair. On the one hand, we have very little definitive knowledge about how to prolong your healthy years aside from the very obvious (exercise, don’t smoke, don’t be fat); on the other hand, by the time we have established that knowledge with much certainty, you may very well no longer be alive to take advantage of it. Coming up with actionable insights is therefore a complex exercise in scientific literacy and fuzzy evaluation of risk-reward tradeoffs. In this post, I would like to describe my own personal thought process and the conclusions to which I’ve come regarding my own “longevity stack.”<\/p>\n\n\n\n

The purpose of this article is not so much to supply a laundry list of supplements but to instead provide the basis of a theoretical framework which the reader may apply to their individual practice. Generally speaking, I do not really like much of what circulates in the wild; people give too much credence to shoddy science but fail to assign enough trust to the best interventions. The most recent example which comes to mind is Bryan Johnson’s Blueprint,<\/a> which may arguably do more harm than good, even if by its expansive nature does include one or two useful treatments.<\/p>\n\n\n\n

The reader should note that this is not meant to be a comprehensive, perfectly reasoned review of all available evidence, which could easily stretch into the hundreds of pages depending on the level of detail demanded. Topics will be covered to the arbitrary level of depth which I personally judge to be appropriate; if felt inadequate, the reader may perform their own research, hopefully using the citations in this post as a useful springboard. Ultimately, if we want to apply this research to our own lives, we must accept some degree of inherent uncertainty, which the structure of this post intentionally reflects.<\/p>\n\n\n\n

Disclaimer: This web site is provided for educational and informational purposes only and does not constitute providing medical advice or professional services. The information provided should not be used for diagnosing or treating a health problem or disease, and those seeking personal medical advice should consult with a licensed physician. <\/em><\/p>\n\n\n\n

General approach<\/strong><\/p>\n\n\n\n

Let’s think a little bit about what the best piece of evidence for a given “anti-aging” treatment would be. In an ideal world, we would have evidence from multiple randomized interventional studies performed on large, representative, healthy human samples that independently demonstrate reproducible lifespan extension and delay of average onset for multiple age-related diseases. Obviously, if any such evidence existed for some compound, we would all know about it, and most of us would already be taking it; needless to say, there is no intervention whatsoever for which this level of evidence exists, even for interventions that “we all know” are good for us, like routine exercise.<\/p>\n\n\n\n

We can weaken this requirement in two primary ways. First, we can accept evidence from nonhumans, allowing us to consider evidence generated from model organisms such as mice, C. elegans,<\/em> and yeast. Such research may not translate cleanly to humans (if at all), but it stands to reason that there must exist mechanisms for extending lifespan which are conserved across species. Second, we can accept noninterventional human evidence, sacrificing some ability to determine causality but allowing us to combine compelling cross-sectional data with theoretical understanding of plausible anti-aging mechanisms. We will see that the most interesting therapeutics by and large tend to fall into one of these two categories.<\/p>\n\n\n\n

Not all evidence is created equal. A core principle of my process of evaluation is that well over 50% of modern biological research is literally fake (fabricated from whole cloth), essentially fake (p<\/em>-hacked or otherwise selectively presented), or so incompetently executed as to be pure noise. Additionally, entire fields of research are contaminated by severe financial conflicts of interest, and all academic research is governed by incentive structures hardly conducive to determination of absolute truth. Unfortunately, we only live once, and it is much easier to harm a complex physiological system than to improve it, so it seems prudent to apply a fairly conservative filter on the set of interventions that we choose to apply to our own bodies.<\/p>\n\n\n\n

Additionally, concerns about drug-drug interactions, as well as the fact that known interventions likely target overlapping mechanisms (leading to rapidly diminishing returns if treatments are not carefully selected), are strong motivations to only consider the strongest bodies of evidence rather than adopting a “bucket list” approach. It is better to properly target a small number of known mechanisms in the safest ways possible using highly specific drugs than to take a hundred supplements, each with weak but pleiotropic effects and unknown absorption profiles, and hope that a good outcome is somehow achieved.<\/p>\n\n\n\n

Overall, we are faced with a problem of considerable difficulty. Nevertheless, I believe that we now understand enough about human physiology and the effects of various drugs that a number of actionable recommendations can be produced from the mists of biomedical research. Let’s take a look.<\/p>\n\n\n\n

Evidence from model organisms: the NIA ITP<\/strong><\/p>\n\n\n\n

Among model organisms, the most well-understood mammal is surely the mouse, and the gold standard for lifespan extension in mice is the National Institute of Aging’s Interventions Testing Program (ITP).<\/a> This program tests a small number of drugs in large, well-controlled, multi-center studies to see if they extend the lifespan of genetically heterogeneous mice. <\/p>\n\n\n\n

Several characteristics of the ITP are highly appealing. It uses a high sample size and experiments are well-powered to detect differences in lifespan; it actually measures total mouse lifespan, the most robust endpoint conceivable and one for which differences have the highest probability of translating to other species; it is a multi-center study performed in standardized conditions where control mice are not subject to artificially adverse environments; treatments are preregistered so results are not p<\/em>-hacked; finally, the mice used are all from the standardized UM-HET3 four-way cross which do not have the genetic peculiarities of the vastly more common and severely inbred C57BL\/6 strain. In short, it is a good experiment. On top of this, the ITP is an expensive program, so the set of compounds tested is selected based on the strength of preexisting evidence, meaning that there is typically a large body of supporting literature to corroborate positive results.<\/p>\n\n\n\n

It is not uncommon for interventions to show lifespan extension in a lower-quality mouse study and to then fail to replicate in the ITP. The reasons why vary, but typically this is because a highly inbred strain is used in preliminary studies or because the mice are not housed properly (control group shows depressed lifespan); in both cases, the treatment is essentially fixing something that is deeply broken, rather than showing a benefit on top of a healthy baseline. For example, one might think that Dang et al.<\/em> (2019)<\/a> supplies intriguing evidence for pro-longevity effects of berberine, until one notices that the median lifespan in both control and treatment groups are below 700 days. We know that properly cared for C57BL\/6 mice have a median lifespan of ~850 days,<\/a> suggesting that the effects of berberine may not replicate in the context of a less dysfunctional colony.<\/p>\n\n\n\n

If we want to choose treatments with the strongest evidence in model organisms, then, a logical place to look is at the ITP’s experimental results.<\/a> In rough order of most to least replication within the ITP itself, the following compounds have been shown to extend both median and maximum lifespan: rapamycin, acarbose, canagliflozin, 17\u03b1-estradiol (males), and glycine (males only; weak effect), as well as various combinations thereof. (Metformin also extended lifespan, but only in combination with rapamycin; this is discussed further in a subsequent section.) The evidence for rapamycin is especially strong, with clear extension of lifespan by 20% or more across different starting ages and treatment regimes, suggesting that it is one of our strongest candidates for lifespan-extending treatments.<\/p>\n\n\n\n

The case for rapamycin<\/strong><\/p>\n\n\n\n

The literature on rapamycin and its ability to extend lifespan at the correct dosages is extensive, so we will only briefly touch on its history here; much more detail is given in the magisterial review by Mannick and Lamming (2023).<\/a> Briefly, genetic inhibition of the mTORC1 kinase was found more than two decades ago to extend the lifespan of multiple model organisms, a result which was then replicated many times over with rapamycin, an orally available inhibitor of mTORC1. At high doses, rapamycin is used as a potent immunosuppressant that prevents post-transplant rejection; however, evidence from animal models suggests that intermittent or low-dose treatment prolongs lifespan without exposure to undesirable side effects, such as total immunosuppression, hyperlipidemia, or hyperglycemia.<\/p>\n\n\n\n

Rapamycin treatment seems to extend lifespan under a variety of different conditions. For example, Miller et al.<\/em> (2014)<\/a> and Miller et al.<\/em> (2010)<\/a> also report that the greatest rapamycin-dependent lifespan extension in the ITP is seen with treatment starting at 9 months of age in mice (equivalent to a 30-year-old human), with 23% extension in males and 26% in females:<\/p>\n\n\n

\n
\"\"<\/figure><\/div>\n\n\n

However, even when treatment was started in extremely old age (Harrison et al.<\/em> (2009))<\/a>, rapamycin extended lifespan by 9% in males and 14% in females:<\/p>\n\n\n

\n
\"\"<\/figure><\/div>\n\n\n

A starting age of 600 days in mice corresponds to approximately 60 years of age in humans, suggesting that a substantial benefit can be seen even if treatment is not started until one’s later years. (Note that the above graph demonstrates an artefactual separation of the survival curve for male mice even before the 600-day mark; the authors attribute this to unusual conditions at one or two specific study sites.)<\/p>\n\n\n\n

Effects are also detected across varying treatment regimes, e.g.<\/em> single dose vs. intermittent vs. continual administration (see for example Bitto et al.<\/em> (2016)<\/a> on transient usage in middle-aged mice), and rapamycin’s ability to extend lifespan is additive when combined with mechanistically distinct drugs like acarbose. Overall, the robustness of these results suggest that the beneficial effects of rapamycin have a relatively higher likelihood of translating to humans.<\/p>\n\n\n\n

The fact that rapamycin is clinically used (at very high doses, achieving serum concentrations much greater than those of the mice in the studies above) as an immunosuppressant means that a priori,<\/em> the most concerning potential side effect of rapamycin treatment, even at lower doses, is undesirable immune suppression. Curiously, however, limited evidence from human trials suggests that mTORC1 inhibition in the elderly can actually improve<\/em> immune function:<\/p>\n\n\n\n